WO2000049150A1 - Constructions geniques permettant de produire un poisson d'agrement transgenique fluorescent - Google Patents

Constructions geniques permettant de produire un poisson d'agrement transgenique fluorescent Download PDF

Info

Publication number
WO2000049150A1
WO2000049150A1 PCT/SG1999/000079 SG9900079W WO0049150A1 WO 2000049150 A1 WO2000049150 A1 WO 2000049150A1 SG 9900079 W SG9900079 W SG 9900079W WO 0049150 A1 WO0049150 A1 WO 0049150A1
Authority
WO
WIPO (PCT)
Prior art keywords
fish
gene
gly
leu
glu
Prior art date
Application number
PCT/SG1999/000079
Other languages
English (en)
Inventor
Zhiyuan Gong
Toong Jin Lam
Bensheng Ju
Original Assignee
National University Of Singapore
Xu, Yianfei
He, Jiangyan
Yan, Tie
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National University Of Singapore, Xu, Yianfei, He, Jiangyan, Yan, Tie filed Critical National University Of Singapore
Priority to US09/913,898 priority Critical patent/US7135613B1/en
Publication of WO2000049150A1 publication Critical patent/WO2000049150A1/fr
Priority to US10/605,708 priority patent/US7834239B2/en
Priority to US11/749,032 priority patent/US8153858B2/en
Priority to US13/334,444 priority patent/US8378169B2/en
Priority to US13/738,704 priority patent/US9763430B2/en
Priority to US15/674,798 priority patent/US11259509B2/en
Priority to US17/680,843 priority patent/US20220256819A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43595Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from coelenteratae, e.g. medusae
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/20Animal model comprising regulated expression system
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/40Fish
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/02Animal zootechnically ameliorated
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0393Animal model comprising a reporter system for screening tests
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/42Vector systems having a special element relevant for transcription being an intron or intervening sequence for splicing and/or stability of RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron

Definitions

  • This invention relates to fish gene promoters and chimeric gene constructs with these promoters for generation of transgenic fish, particularly fluorescent transgenic ornamental fish.
  • Transgenic technology involves the transfer of a foreign gene into a host organism enabling the host to acquire a new and inheritable trait.
  • the technique was first developed in mice by Gordon et al. (1980). They injected foreign DNA into fertilized eggs and found that some of the mice developed from the injected eggs retained the foreign DNA. Applying the same technique, Palmiter et al. (1982) have introduced a chimeric gene containing a rat growth hormone gene under a mouse heavy metal-inducible gene promoter and generated the first batch of genetically engineered supermice, which are almost twice as large as non-transgenic siblings. This work has opened a promising avenue in using the transgenic approach to provide to animals new and beneficial traits for livestock husbandry and aquaculture.
  • transgenic technology In addition to the stimulation of somatic growth for increasing the gross production of animal husbandry and aquaculture, transgenic technology also has many other potential applications.
  • transgenic animals can be used as bioreactors to produce commercially useful compounds by expression of a useful foreign gene in milk or in blood.
  • Many pharmaceutically useful protein factors have been expressed in this way.
  • human ⁇ l-antitrypsin which is commonly used to treat emphysema, has been expressed at a concentration as high as 35 mg/ml (10% of milk proteins) in the milk of transgenic sheep (Wright et al., 1991).
  • transgenic technique can also be used to improve the nutritional value of milk by selectively increasing the levels of certain valuable proteins such as caseins and by supplementing certain new and useful proteins such as lysozyme for antimicrobial activity (Maga and Murray, 1995).
  • transgenic mice have been widely used in medical research, particularly in the generation of transgenic animal models for human disease studies (Lathe and Mullins, 1993). More recently, it has been proposed to use transgenic pigs as organ donors for xenotransplantation by expressing human regulators of complement activation to prevent hyperacute rejection during organ transplantation (Cozzi and White, 1995). The development of disease resistant animals has also been tested in transgenic mice (e.g. Chen et al, 1988).
  • the zebrafish Danio rerio
  • the zebrafish is a new model organism for vertebrate developmental biology.
  • the zebrafish offers several major advantages such as easy availability of eggs and embryos, tissue clarity throughout embryogenesis, external development, short generation time and easy maintenance of both the adult and the young.
  • Transgenic zebrafish have been used as an experimental tool in zebrafish developmental biology.
  • GFP green fluorescent protein
  • the wild type GFP emits green fluorescence at a wavelength of 508 nm upon stimulation with ultraviolet light (395 nm).
  • the primary structure of GFP has been elucidated by cloning of its cD ⁇ A and genomic D ⁇ A (Prasher et al., 1992).
  • a modified GFP, also called EGFP (Enhanced Green Fluorescent Protein) has been generated artificially and it contains mutations that allow the protein to emit a stronger green light and its coding sequence has also been optimized for higher expression in mammalian cells based on preferable human codons.
  • EGFP fluorescence is about 40 times stronger than the wild type GFP in mammalian cells (Yang et al., 1996).
  • GFP (including EGFP) has become a popular tool in cell biology and transgenic research.
  • the GFP fusion-protein can be used as an indicator of the subcellular location of the tested protein (Wang and Hazelrigg, 1994) .
  • the GFP can be used as a marker to identify expressing cells (Chalfie et al., 1994).
  • the GFP gene has become an increasingly popular reporter gene for transgenic research as GFP can be easily detected by a non-invasive approach.
  • the GFP gene (including EGFP gene) has also been introduced into zebrafish in several previous reports by using various gene promoters, including Xenopus elongation factor la enhancer-promoter (Amsterdam et al., 1995, 1996), rat myosin light-chain enhancer (Moss et al., 1996), zebrafish GATA-1 and GATA-3 promoters (Meng et al., 1997;
  • zebrafish gene promoters of different characteristics were isolated and four chimeric gene constructs containing a zebrafish gene promoter and EGFP DNA were made: pCK-EGFP, pMCK-EGFP, pMLC2f-EGFP and pARP-EGFP.
  • the first chimeric gene-construct, pCK-EGFP contains a 2.2 kbp polynucleotide comprising a zebrafish cytokeratin (CK) gene promoter which is specifically or predominantly expressed in skin epithelia.
  • CK zebrafish cytokeratin
  • the second one contains a 1.5 kbp polynucleotide comprising a muscle-specific promoter from a zebrafish muscle creatine kinase (MCK) gene and the gene is only expressed in the muscle tissue.
  • the third construct, pMLC2f-EGFP contains a 2.2 kpb polynucleotide comprising a strong skeletal muscle-specific promoter from the fast skeletal muscle isoform of the myosin light chain 2 (MLC2f) gene and is expressed specifically or predominantly in skeletal muscle.
  • the fourth chimeric gene construct, pARP-EGFP contains a strong and ubiquitously expressed promoter from a zebrafish acidic ribosomal protein (ARP) gene.
  • ARP zebrafish acidic ribosomal protein
  • the GFP transgenic fish emit green fluorescence light under a blue or ultraviolet light and this feature makes the genetically engineered fish unique and attractive in the ornamental fish market.
  • the fluorescent transgenic fish are also useful for the development of a biosensor system and as research models for embryonic studies such as cell lineage, cell migration, cell and nuclear transplantation etc.
  • Figs. 1A-1I are photographs showing expression of CK (Figs. 1A-1C), MCK (Figs. ID-IE), ARP (Figs. 1F-1G) and MLC2f (Figs. 1H-1I) mRNAs in zebrafish embryos as revealed by whole mount in situ hybridization (detailed description of the procedure can be found in Thisse et al., 1993).
  • Fig. 1A A 28 hpf (hour postfertilization) embryo hybridized with a CK antisense riboprobe.
  • Fig. IB Enlargement of the mid-part of the embryo shown in Fig. 1A.
  • Fig. 1C Cross-section of the embryo in Fig. 1A.
  • FIG. IF A 30 hpf embryo hybridized with an MCK antisense riboprobe.
  • Fig. IE Cross-section of the embryo in Fig ID.
  • Fig. IF A 28 hpf embryo hybridized with an ARP antisense riboprobe.
  • Fig. 1G Cross-section of the embryo in Fig. IF. Arrows indicate the planes for cross- sections and the box in panel A indicates the enlarged region shown in panel B.
  • Fig. 1H Side view of a 22-hpf embryo hybridized with the MLC2f probe.
  • Fig. II Transverse section through the trunk of a stained 24-hpf embryo. SC, spinal cord; N, notochord.
  • Fig. 2A is a digitized image showing distribution of CK, MCK and ARP mRNAs in aduk tissues.
  • Total RNAs were prepared from selected adult tissues as indicated at the top of each lane and analyzed by Northern blot hybridization (detailed description of the procedure can be found in Gong et al., 1992). Three identical blots were made from the same set of RNAs and hybridized with the CK, MCK and ARP probes, respectively.
  • Fig. 2B is a digitized image showing distribution of MLC2f mRNA in adult tissues.
  • Total RNAs were prepared from selected adult tissues as indicated at the top of each lane and analyzed by Northern blot hybridization (detailed description of the procedure can be found in Gong et al., 1992). Two identical blots were made from the same set of RNAs and hybridized with the MLC2f probe and a ubiquitously expressed U-actin probe, respectively.
  • Fig. 3. is a schematic representation of the strategy of promoter cloning. Restriction enzyme digested genomic DNA was ligated with a short linker DNA which consists of Oligo 1 and Oligo 2. Nested PCR reactions were then performed: the first round PCR used linker specific primer LI and gene specific primers Gl, where Gl is CK1, MCK1, Ml or ARP1 in the described embodiments, and the second round linker specific primer L2 and gene specific primer G2, where G2 is CK2, MCK2, M2 or ARP2, respectively in the described embodiments.
  • Fig. 4 is a schematic map of the chimeric gene construct, pCK-EGFP.
  • the 2.2 kb zebrafish DNA fragment comprising the CK promoter region is inserted into pEGFP-1 (Clonetech) at the EcoRI and BamHI site as indicated.
  • the EGFP gene is under control of the zebrafish CK promoter.
  • the kanamycin neomycin resistance gene (Karf/Neo 1 ) in the backbone of the original pEGFP-1 plasmid.
  • the total length of the recombinant plasmid pCK-EGFP is 6.4 kb.
  • Fig. 5 is a schematic map of the chimeric gene construct, pMCK-EGFP.
  • the 1.5 kb zebrafish DNA fragment comprising the MCK promoter region is inserted into pEGFP-1 (Clonetech) at the EcoRI and BamHI site as indicated.
  • the EGFP gene is under control of the zebrafish MCK promoter.
  • the kanamycin/neomycin resistance gene Kan r /Neo r
  • the total length of the recombinant plasmid pMCK-EGFP is 5.7 kb.
  • Fig. 6 is a schematic map of the chimeric gene construct, pARP-EGFP.
  • the 2.2 kb zebrafish DNA fragment comprising the ARP promoter/1 st intron region is inserted into pEGFP-1 (Clonetech) at the EcoRI and BamHI site as indicated.
  • the EGFP gene is under control of the zebrafish ARP promoter.
  • the kanamycin/neomycin resistance gene Kan r /Neo r
  • the total length of the recombinant plasmid pARP-EGFP is 6.4 kb. -
  • Fig. 7 is a schematic map of the chimeric gene construct, pMLC2f-EGFP.
  • the 2.0 kb zebrafish DNA fragment comprising the MLC2f promoter region is inserted into pEGFP-1 (Clonetech) at the Hindlll and BamHI site as indicated.
  • the EGFP gene is under control of the zebrafish MLC2f promoter.
  • the kanamycin/neomycin resistance gene Kan r /Neo r
  • the total length of the recombinant plasmid pMLC2f-EGFP is 6.2 kb.
  • Fig. 8 is a photograph of a typical transgenic zebrafish fry (4 days old) with pCK- EGFP, which emits green fluorescence from skin epithelia under a blue light.
  • Fig. 9 is a photograph of a typical transgenic zebrafish fry (3 days old) with pMCK- EGFP, which emits green fluorescence from skeletal muscles under a blue light.
  • Fig. 10 is a photograph of a typical transgenic zebrafish fry (2 days old) with pARP-EGFP, which emits green fluorescence under a blue light from a variety of cell types such as skin epithelia, muscle cells, lens, neural tissues, notochord, circulating blood cells and yolk cells.
  • Figs. 11A-11B Photographs of a typical transgenic zebrafish founder with pMLC2f-EGFP (Fig. 11 A) and an FI stable transgenic offspring (Fig. 1 IB). Both pictures were taken under an ultraviolet light (365 nm). The green fluorescence can be better observed under a blue light with an optimal wavelength of 488 nm.
  • Figs. 12A-12C Examples of high, moderate and low expression of GFP in transiently transgenic embryos at 72 hpf.
  • Fig. 12 A High expression, GFP expression was detected in essentially 100% of the muscle fibers in the trunk.
  • Fig. 12B Moderate expression, GFP expression was detected in several bundles of muscle fibers, usually in the mid-trunk region.
  • Fig. 12C Low expression, GFP expression occurred in dispersed muscle fibers and the number of GFP positive fibers is usually less than 20 per embryo.
  • Fig. 13 Deletion analysis of the MLC2f promoter in transient transgenic zebrafish embryos.
  • a series of 5' deletions of MLC2f-EGFP constructs containing -2011-bp (2-kb), -1338-bp, -873-bp, -283-bp, -77-bp and -3-bp of the MLC2f promoter were generated by unidirectional deletion using the double-stranded Nested Deletion Kit from Pharmacia based on the manufacturer's instructional manual.
  • Each construct was injected into approximately 100 embryos and GFP expression was monitored in the first 72 hours of embryonic development. The level of GFP expression was classified based on the examples shown in Figs. 12A-12C.
  • Potential E-boxes and MEF2 binding sites, which are important for muscle-specific transcription are indicated on the -2011-bp construct.
  • the gene construct generally comprises three portions: a gene promoter, a structural gene and transcriptional termination signals.
  • the gene promoter would determine where, when and under what conditions the structural gene is turned on.
  • the structural gene contains protein coding portions that determine the protein to be synthesized and thus the biological function.
  • the structural gene might also contain intron sequences which can affect mRNA stability or which might contain transcription regulatory elements.
  • the transcription termination signals consist of two parts: a polyadenylation signal and a transcriptional termination signal after the polyadenylation signal. Both are important to terminate the transcription of the gene.
  • selection of a promoter is very important for successful transgenic study, and it is preferable to use a homologous promoter (homologous to the host fish) to ensure accurate gene activation in the transgenic host.
  • a promoter drives expression "predominantly” in a tissue if expression is at least 2- fold, preferably at least 5-fold higher in that tissue compared to a reference tissue.
  • a promoter drives expression "specifically” in a tissue if the level of expression is at least 5- fold, preferably at least 10-fold higher, more preferably at least 50-fold higher in that tissue than in any other tissue.
  • Recombinant DNA constructs comprising one or more of the DNA or RNA sequences described herein and an additional DNA and/or RNA sequence are also included within the scope of this invention. These recombinant DNA constructs usually have sequences which do not occur in nature or exist in a form that does not occur in nature or exist in association with other materials that do not occur in nature.
  • the DNA and/or R A sequences described as constructs or in vectors above are "operably linked" with other DNA and/or RNA sequences. DNA regions are operably linked when they are functionally related to each other.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as part of a preprotein which participates in the-secretion of the polypeptide; a promoter is operably linked to a coding sequence if it controls the transcription of the coding sequence; a ribosome binding site is operably linked to a coding sequence if it is positioned so as to permit translation.
  • operably linked means contiguous (or in close proximity to) and, in the case of secretory leaders, contiguous and in reading phase.
  • CK cytokeratin
  • SEQ ID NO:l The complete cytokeratin (CK) cDNA sequence is shown in SEQ ID NO:l, and its deduced amino acid sequence is shown in SEQ ID NO:2.
  • the extra nucleotides introduced into CK2 for generation of a restriction site are shown as a miscjfeature in the primer sequence SEQ ID NO:l l.
  • a potential polyadenylation signal, AATAAA is indicated in SEQ ID NO:l.
  • MCK muscle creatine kinase
  • SEQ ID NO:3 The complete muscle creatine kinase (MCK) cDNA sequence is shown in SEQ ID NO:3, and its deduced amino acid sequence is shown in SEQ ID NO:4.
  • the binding sites of the gene specific primers for promoter amplification, MCKl and MCK2, are indicated.
  • the extra nucleotides introduced into MCKl and MCK2 for generation of restriction sites are shown as a misc_feature in the primer sequences SEQ ID NOS: 12 and 13, respectively.
  • a potential polyadenylation signal, AATAAA is indicated in SEQ ID NO:3.
  • the complete fast skeletal muscle isoform of myosin light chain 2 (MLC2f) cDNA sequence is shown in SEQ ID NO:20, and its deduced amino acid sequence is shown in SEQ ID NO:21.
  • Ml and M2 are indicated.
  • Two potential polyadenylation signals, AATAAA, are shown as a misc_feature in SEQ ID NO:20.
  • ARP ribosomal protein P0 cDNA sequence
  • SEQ ID NO:5 The complete acidic ribosomal protein P0 (ARP) cDNA sequence is shown in SEQ ID NO:5, and its deduced amino acid sequence is shown in SEQ ID NO:6.
  • the extra nucleotides introduced into ARP2 for generation of a restriction site are shown as a misc_feature in the primer sequence SEQ ID NO: 15.
  • a potential polyadenylation signal, AATAAA is indicated in SEQ ID NO:5.
  • SEQ ID NO:7 shows the complete sequence of the CK promoter region.
  • a putative TATA box is shown, and the 3' nucleotides identical to the 5' CK cDNA sequence are shown as a misc_feature.
  • the binding site of the second gene specific primer, CK2 is shown.
  • the introduced BamHI site is indicated as a misc_feature in the primer sequence SEQJD NO:l l.
  • SEQ ID NO:8 shows the complete sequence of the MCK promoter region. A putative TATA box is shown, and the 3' nucleotides identical to the 5' MCK cDNA sequence are shown as a misc feature in SEQ ID NO: 8. The binding site of the second gene specific primer, MCK2, is shown. The introduced BamHI site is indicated as a misc_feature in the primer sequence SEQ ID NO: 13.
  • SEQ ID NO:22 shows the complete sequence of the MLC2f promoter region.
  • a putative TATA box is shown, and the 3' nucleotides identical to the 5' MLC2f cDNA sequence are shown as a misc eature.
  • the binding site of the second gene-specific primer, M2 is shown. Potential muscle-specific cis-elements, E-boxes and MEF2 binding sites, are also shown.
  • the proximal 1-kb region of the MLC2f promoter was recently published (Xu et al., 1999).
  • SEQ ID NO: 9 shows the complete sequence of the ARP promoter region including the first intron.
  • the first intron is shown, and the 3' nucleotides identical to the 5' ARP cDNA sequence are shown as misc_features. No typical TATA box is found.
  • the binding site of the second gene specific primer, ARP2 is shown.
  • the introduced BamHI site is indicated as a misc_feature in the primer sequence SEQ ID NO : 15.
  • the present invention contemplates use of DNA that codes for various polypeptides and other types of DNA to prepare the gene constructs of the present invention.
  • DNA that codes for structural proteins such as fluorescent peptides including GFP, EGFP, BFP, EBFP, YFP, EYFP, CFP, ECFP and enzymes (such as luciferase, ⁇ -galactosidase, chloramphenicol acetyltransferase, etc.), and hormones (such as growth hormone etc.), are useful in the present invention. More particularly, the DNA may code for polypeptides comprising the sequences exemplified in SEQ ID NOS:2, 4, 6 and 21.
  • the present invention also contemplates use of particular DNA sequences, including regulatory sequences, such as promoter sequences shown in SEQ ID NOS: 7, 8, 9 and 22 or portions thereof effective as promoters. Finally, the present invention also contemplates the use of additional DNA sequences, described generally herein or described in the references cited herein, for various purposes. Chimeric Genes
  • the present invention also encompasses chimeric genes comprising a promoter described herein operatively linked to a heterologous gene.
  • a chimeric gene can comprise a promoter of a zebrafish operatively linked to a zebrafish structural gene other than that normally found linked to the promoter in the genome.
  • the promoter can be operatively linked to a gene that is exogenous to a zebrafish, as exemplified by the GFP and other genes specifically exemplified herein.
  • a chimeric gene can comprise an exogenous promoter linked to any structural gene not normally linked to that promoter in the genome of an organism.
  • DNA that codes for variants of the specifically exemplified polypeptides are also encompassed by the present invention.
  • Possible variants include allelic variants and corresponding polypeptides from other organisms, particularly other organisms of the same species, genus or family.
  • the variants may have substantially the same characteristics as the natural polypeptides.
  • the variant polypeptide will possess the primary property of concern for the polypeptide.
  • the polypeptide will possess one or more or all of the primary physical (e.g., solubility) and/or biological (e.g., enzymatic activity, physiologic activity or fluorescence excitation or emission spectrum) properties of the reference polypeptide.
  • DNA of the structural genes of the present invention will encode a protein that produces a fluorescent or chemiluminescent light under conditions appropriate to the particular polypeptide in one or more tissues of a fish.
  • Preferred tissues for expression are skin, muscle, eye and bone.
  • the polypeptide may have additional individual amino acids or amino acid sequences inserted into the polypeptide in the middle thereof and/or at the N-terminal and/or C-terminal ends thereof so long as the polypeptide possesses the desired physical and/or biological characteristics.
  • some of the amino acids or amino acid sequences may be deleted from the polypeptide so long as the polypeptide possesses the desired physical and/or biochemical characteristics. Amino acid substitutions may also be made in the sequences so long as the polypeptide possesses the desired physical and biochemical characteristics.
  • DNA coding for these variants can be used to prepare gene constructs of the present invention. Sequence Identity
  • variants of polypeptides or polynucleotides contemplated herein should possess more than 75% sequence identity (sometimes referred to as homology), preferably more than 85% identity, most preferably more than 95% identity, even more preferably more than 98% identity to the naturally occurring and/or specifically exemplified sequences or fragments thereof described herein. To determine this homology, two sequences are aligned so as to obtain a maximum match using gaps and inserts.
  • sequences are said to be “identical” if the sequence of residues is the same when aligned for maximum correspondence as described below.
  • complementary applies to nucleic acid sequences and is used herein to mean that the sequence is complementary to all or a portion of a reference polynucleotide sequence.
  • Optimal alignment of sequences for comparison can be conducted by the local homology algorithm of Smith and Waterman (1981), by the homology alignment method of Needleman and Wunsch (1970), by the search for similarity method of Pearson and Lippman (1988), or the like.
  • Computer implementations of the above algorithms are known as part of the Genetics Computer Group (GCG) Wisconsin Genetics Software Package (GAP, BESTFIT, BLASTA, FASTA and TFASTA), 575 Science Drive, Madison, WI. These programs are preferably run using default values for all parameters.
  • Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the sequence in the comparison window may comprise additions or deletions (i.e. "gaps") as compared to the reference sequence for optimal alignment of the two sequences being compared.
  • the percentage identity is calculated by determining the number of positions at which the identical residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window and multiplying the result by 100 to yield the percentage of sequence identity. Total identity is then determined as the average identity over all of the windows that cover the complete query sequence.
  • the invention encompasses DNA that codes for any one of the above-described polypeptides including, but not limited to, those shown in SEQ ID NOS:2, 4, 6 and 21 including fusion polypeptides, variants and fragments thereof.
  • the sequence of certain particularly useful cDNAs which encode polypeptides are shown in SEQ ID NOS:l, 3, 5 and 20.
  • the present invention also includes cDNA as well as genomic DNA containing or comprising the requisite nucleotide sequences as well as corresponding RNA and antisense sequences.
  • allelic variants are a sequence that is a variant from that of the exemplified nucleotide sequence, but represents the same chromosomal locus in the organism.
  • allelic variants can be produced by genetic engineering methods.
  • a preferred allelic variant is one that is found in a naturally occurring organism, including a laboratory strain.
  • Allelic variants are either silent or expressed.
  • a silent allele is one that does not affect the phenotype of the organism. An expressed allele results in a detectable change in the phenotype of the trait represented by the locus.
  • a nucleic acid sequence "encodes” or “codes for” a polypeptide if it directs the expression of the polypeptide referred to.
  • the nucleic acid can be DNA or RNA.
  • a nucleic acid sequence that encodes a polypeptide includes the transcribed strand, the hnRNA and the spliced RNA or the DNA representative of the mRNA.
  • An "antisense” nucleic acid is one that is complementary to all or part of a strand representative of mRNA, including untranslated portions thereof.
  • the DNA of the present invention may also have any base sequence that has been changed by substitution in accordance with degeneracy of genetic code.
  • the DNA is readily modified by substitution, deletion or insertion of nucleotides, thereby resulting in novel DNA sequences encoding the polypeptide or its derivatives. These modified sequences are used to produce mutant polypeptide and to directly express the polypeptide. Methods for saturating a particular DNA sequence with random mutations and also for making specific site-directed mutations are known in the art; see e.g. Sambrook et al. (1989). Hybridizable Variants
  • the DNA molecules useful in accordance with the present invention can comprise a nucleotide sequence selected from the group consisting of SEQ ID NOS.:l, 3, 5, 7-20 and 22-24 or can comprise a nucleotide sequence that hybridizes to a DNA molecule comprising the nucleotide sequence of SEQ ID NOS.:l, 3, 5 or 20 under salt and temperature conditions providing stringency at least as high as that equivalent to 5x SSC and 42°C and that codes on expression for a polypeptide that has one or more or all of the above-described physical and/or biological properties.
  • the present invention also includes polypeptides coded for by these hybridizable variants.
  • the present invention also encompasses functional promoters which hybridize to SEQ ID NOS:7, 8, 9 or 22 under the above- described conditions. DNA molecules of the invention will preferably hybridize to reference sequences under more stringent conditions allowing the degree of mismatch represented by the degrees of sequence identity enumerated above.
  • the present invention also encompasses functional primers or linker oligonucleotides set forth in SEQ ID NOS:10-19 and 23-24 or larger primers comprising these sequences, or sequences which hybridize with these sequences under the above-described conditions.
  • the primers usually have a length of 10-50 nucleotides, preferably 15-35 nucleotides, more preferably 18-30 nucleotides.
  • the invention is further directed to a replicable vector containing cDNA that codes for the polypeptide and that is capable of expressing the polypeptide.
  • the present invention is also directed to a vector comprising a replicable vector and a DNA sequence corresponding to the above described gene inserted into said vector.
  • the vector may be an integrating or non-integrating vector depending on its intended use and is conveniently a plasmid.
  • the invention further relates to a transformed cell or microorganism containing cDNA or a vector which codes for the polypeptide or a fragment or variant thereof and that is capable of expressing the polypeptide.
  • vertebrate host cell lines useful in the present invention preferably include cells from any of the fish described herein.
  • Expression vectors for such cells ordinarily include (if necessary) an origin of replication, a promoter located upstream from the gene to be expressed, along with a ribosome-binding site, RNA splice site (if intron-containing genomic DNA is used or if an intron is necessary to optimize expression of a cDNA), a polyadenylation site, and a transcription termination sequence.
  • Example I Isolation of skin-specific, muscle-specific and ubiquitously expressed zebrafish cDNA clones.
  • cDNA clones were isolated and sequenced as described by Gong et al. (1997). Basically, random cDNA clones were selected from zebrafish embryonic and adult cDNA libraries and each clone was partially sequenced by a single sequencing reaction. The partial sequences were then used to identify the sequenced clones for potential function and tissue specificity.
  • clones identified by this approach four of them were selected: for skin specificity (clone A39 encoding cytokeratin, CK), for muscle specificity (clone El 46 encoding muscle creatine kinase, MCK), for skeletal muscle specificity (clone A113 encoding the fast skeletal muscle isoform of the myosin light chain 2, MLC2f) and for ubiquitous expression (clone A150 encoding acidic ribosomal protein P0, ARP), respectively.
  • skin specificity clone A39 encoding cytokeratin, CK
  • muscle specificity clone El 46 encoding muscle creatine kinase, MCK
  • skeletal muscle specificity clone A113 encoding the fast skeletal muscle isoform of the myosin light chain 2, MLC2f
  • ubiquitous expression clone A150 encoding acidic ribosomal protein P0, ARP
  • the four cDNA clones were sequenced, and their complete cDNA sequences with deduced amino acid sequences are shown in SEQ ID NOS:l, 3, 5, and 20 respectively.
  • A39 encodes a type II basic cytokeratin and its closest homolog in mammals is cytokeratin 8 (65-68% amino acid identity).
  • E146 codes for the zebrafish MCK and its amino acid sequence shares ⁇ 87% identity with mammalian MCKs.
  • A113 encodes the fast skeletal muscle isoform of the myosin light chain 2.
  • the deduced amino acid sequence of this gene is highly homologous to other vertebrate fast skeletal muscle MLC2f proteins (over 80% amino acid identity).
  • the amino acid sequence of zebrafish ARP deduced from the A 150 clone is 87-89% identical to those of mammalian ARPs.
  • cytokeratin mRNA was specifically expressed in the embryonic surface (Figs. 1A-1C ) and cross section of in situ hybridized embryos confirmed that the expression was only in skin epithelia (Fig. 1C).
  • the cytokeratin mRNA appeared before 4 hours post-fertilization (hpf) and it is likely that the transcription of the cytokeratin gene starts at mid-blastula transition when the zygotic genome is activated.
  • hpf post-fertilization
  • a clear cytokeratin mRNA signal was detected in highly flattened cells of the superficial layer in blastula and the expression remained in the superficial layer which eventually developed into skin epithelia including the yolk sac.
  • cytokeratin mRNA In adult tissues, cytokeratin mRNA was predominantly detected in the skin and also weakly in several other tissues including the eye, gill, intestine and muscle, but not in the liver and ovary (Fig. 2). Therefore, the cytokeratin mRNA is predominantly, if not specifically, expressed in skin cells.
  • MCK mRNA was first detected in the first few anterior somites in 10 somite stage embryos (14 hpf) and at later stages the expression is specifically in skeletal muscle (Fig. ID) and in heart (data not shown). When the stained embryos are cross-sectioned, the
  • MCK mRNA signal was found exclusively in the trunk skeletal muscles (Fig. IE). In adult tissues, MCK mRNA was detected exclusively in the skeletal muscle (Fig. 2).
  • MLC2f mRNA was specifically expressed in fast skeletal muscle in developing zebrafish embryos (Figs.1H- II). To examine the tissue distribution of MLC2f mRNA, total RNAs were prepared from several adult tissues including heart, brain, eyes, gills, intestine, liver, skeletal muscle, ovary, skin, and testis. MLC2f mRNA was only detected in the skeletal muscle by Northern analysis; while -actin mRNA was detected ubiquitously in the same set of RNAs, confirming the validity of the assay (Fig. 2B).
  • ARP mRNA was expressed ubiquitously and it is presumably a maternal mRNA since it is present in the ovary as well as in embryos at one cell stage. In in situ hybridization experiments, an intense hybridization signal was detected in most tissues. An example of a hybridized embryo at 28 hpf is shown in Fig. IF. In adults, ARP mRNA was abundantly expressed in all tissues examined except for the brain where a relatively weak signal was detected (Fig. 2A). These observations confirmed that the ARP mRNA is expressed ubiquitously.
  • Example II Isolation of zebrafish gene promoters
  • zebrafish gene promoters were isolated by a linker-mediated PCR method as described by Liao et al., (1997) and as exemplified by the diagrams in Fig. 3.
  • the whole procedure includes the following steps: 1) designing of gene specific primers; 2) isolation of zebrafish genomic DNA; 3) digestion of genomic DNA by a restriction enzyme; 4) ligation of a short linker DNA to the digested genomic DNA; 5) PCR amplification of the promoter region; and 6) DNA sequencing to confirm the cloned DNA fragment. The following is the detailed description of these steps.
  • Gene specific PCR primers were designed based on the 5' end of the four cDNA sequences and the regions used for designing the primers are shown in SEQ ID NOS: 1, 3, 5 and 20.
  • the two cytokeratin gene specific primers are:
  • CK2 (SEQ ID NO:l 1), where the first six nucleotides are for creation of an EcoRI site to facilitate cloning.
  • the two muscle creatine kinase gene specific primers are:
  • MCK2 (SEQ ID NO: 13), where the first three nucleotides are for creation of an EcoRI site to facilitate cloning.
  • the two fast skeletal muscle isoform of myosin light chain 2 gene specific primers are:
  • the two acidic ribosomal protein P0 gene specific primers are:
  • ARPl (SEQ ID NO: 14) ARP2 (SEQ ID NO: 15), where the first six nucleotides are for creation of an EcoRI site to facilitate cloning.
  • Genomic DNA was isolated from a single individual fish by a standard method (Sambrook et al, 1989). Generally, an adult fish was quickly frozen in liquid nitrogen and ground into powder. The ground tissue was then transferred to an extraction buffer (10 mM Tris, pH 8,
  • Genomic DNA was digested with the selected restriction enzymes. Generally, 500 units of restriction enzyme were used to digest 50 ⁇ g of genomic DNA overnight at the optimal enzyme reaction temperature (usually at 37°C).
  • the linker DNA was assembled by annealing equal moles of the two linker oligonucleotides, Oligol (SEQ ID NO:16) and Oligo 2 (SEQ ID NO: 17). Oligo 2 was phosphorylated by T4 polynucleotide kinase prior to annealing. Restriction enzyme digested genomic DNA was filled-in or trimmed with T4 DNA polymerase, if necessary, and ligated with the linker DNA. Ligation was performed with 1 ⁇ g of digested genomic DNA and 0.5 ⁇ g of linker DNA in a 20 ⁇ l reaction containing 10 units of T4 DNA ligase at 4°C overnight.
  • PCR was performed with Advantage Tth Polymerase Mix (Clontech). The first round of PCR was performed using a linker specific primer LI (SEQ ID NO: 18) and a gene specific primer Gl (CK1, MCKl, Ml or ARPl).
  • LI linker specific primer
  • Gl gene specific primer Gl
  • the cycling conditions were as follows: 94°C/1 min, 35 cycles of 94°C/30 sec and 68°C/6 min, and finally 68°C/8 min.
  • the products were diluted 100 fold.
  • One ⁇ l of diluted PCR product was used as template for the second round of PCR (nested PCR) with a second linker specific primer L2 (SEQ ID NO: 19) and a second gene specific primer G2 (CK2, MCK2, M2 or ARP2), as described for the primary PCR but with the following modification:
  • PCR products were purified from the agarose gel following electrophoresis and cloned into a TA vector, pT7BlueTM (Novogen). DNA sequencing was performed by dideoxynucleotide chain termination method using a T7 Sequencing Kit purchased from Pharmacia. Complete sequences of these promoter regions were obtained by automatic sequencing using a dRhodamine Terminator Cycle Sequencing Ready Reaction Kit (Perkin-Elmer) and an ABI 377 automatic sequencing machine.
  • the isolated cytokeratin DNA fragment comprising the gene promoter is 2.2 kb.
  • the 3' proximal region immediately upstream of a portion identical to the 3' part of the CK cDNA sequence there is a putative TATA box perfectly matching to a consensus TATA box sequence.
  • the 164 bp of the 3' region is identical to the 5' UTR (untranslated region) of the cytokeratin cDNA.
  • the isolated fragment was indeed derived from the same gene as the cytokeratin cDNA clone (SEQ ID NO:7).
  • MLC2f a 2 kb region was isolated from the fast skeletal muscle isoform of myosin light chain 2 gene and sequenced completely.
  • the promoter sequence for MLC2f is shown in SEQ ID NO:22.
  • the sequence immediately upstream of the gene specific primer M2 is identical to the 5' UTR of the MLC2f cDNA clone; thus, the amplified DNA fragments are indeed derived from the MLC2f gene.
  • a perfect TATA box was found 30 nucleotides upstream of the transcription start site, which was defined by a primer extension experiment based on Sambrook et al. (1989).
  • 2-kb region comprising the promoter, six E-boxes (CANNTG) and six potential MEF2 binding sites [C/T)TA(T/A)4TA(A/G)] were found and are indicated in SEQ ID NO:22. Both of these cis-element classes are important for muscle specific gene transcription (Schwarz et al., 1993; Olson et al., 1995).
  • a 2.2 kb fragment was amplified for the ARP gene.
  • the isolated zebrafish gene promoters were inserted into the plasmid pEGFP-1
  • Linearized plasmid DNAs at a concentrations of 500 ⁇ g/ml (for pCK-EGFP and pMCK-EGFP) and 100 ⁇ g/ml (for pMLC2f-EGFP) in 0.1 M Tris-HCl (pH 7.6)/0.25% phenol red were injected into the cytoplasm of 1- or 2-cell stage embryos. Because of a high mortality rate, pARP-EGFP was injected at a lower concentration (50 ⁇ g/ml). Each embryo received 300-500 pi of DNA.
  • the injected embryos were reared in autoclaved Holtfreter's solution (0.35% NaCl, 0.01% KC1 and 0.01% CaCl2) supplemented with 1 ⁇ g/ml of methylene blue. Expression of GFP was observed and photographed under a ZEISS Axiovert 25 fluorescence microscope.
  • GFP expression started about 4 hours after injection, which corresponds to the stage of ⁇ 30% epiboly. About 55% of the injected embryos expressed GFP at this stage. The early expression was always in the superficial layer of cells, mimicking endogenous expression of the CK gene as observed by in situ hybridization. At later stages, in all GFP-expressing fish, GFP was found predominantly in skin epithelia. A typical pCK-EGFP transgenic zebrafish fry at 4 days old is shown in Fig. 8.
  • p ARP -EGFP expression of p ARP -EGFP was first observed 4 hours after injection at the 30% epiboly stage. The timing of expression is similar to that of pCK-EGFP-injected embryos. However, unlike the pCK-EGFP transgenic embryos, the GFP expression under the ARP promoter occurred not only in the superficial layer of cells but also in deep layers of cells. In some batches of injected embryos, almost 100% of the injected embryos expressed initially. At later stages when some embryonic cells become overtly differentiated, it was found that the GFP expression occurred essentially in all different types of cells such as skin epithelia, muscle cells, lens, neural tissues, notochord, circulating blood cells and yolk cells (Fig. 10).
  • GFP expression Under the MLC2f promoter, nearly 60% of the embryos expressed GFP. The earliest GFP expression started in trunk skeletal muscles about 19 hours after injection, which corresponds to the stage of 20-somite. Later, the GFP expression also occurred in head skeletal muscles including eye muscles, jaw muscles, gill muscles etc.
  • Transgenic founder zebrafish containing pMLC2f-EGFP emit a strong green fluorescent light under a blue or ultraviolet light (Fig. 11A).
  • transgenic offspring were obtained that also displayed strong green fluorescence (Fig. 1 IB).
  • the level of GFP expression is so high in the transgenic founders and offspring that green fluorescence can be observed when the fish are exposed to sunlight.
  • GFP expression was detected in several bundles of muscle fibers, usually in the mid-trunk region.
  • GFP expression occurred in dispersed muscle fibers and the number of GFP positive fibers is usually less than 20 per embryo.
  • deletion up to -283 bp maintained the GFP expression in skeletal muscles in 100%o of the expressing embryos; however, the level of GFP expression from these deletion constructs varies greatly. Strong expression drops from 23%> to 0%> from the 2-kb (-2011 bp) promoter to the -283-bp promoter. Thus, only two constructs (-2011 bp and -1338 bp) are capable of maintaining the high level of expression and the highest expression was obtained only with the 2-kb promoter, indicating the importance of the promoter region of -1338 bp to -2011 bp for conferring the highest promoter activity.
  • the expression of GFP using pMLC2f-EGFP is much higher than that obtained using the pMCK-EGFP that contains a 1.5 kb of zebrafish MCK promoter.
  • pMCK-EGFP that contains a 1.5 kb of zebrafish MCK promoter.
  • the promoters of the genes represented by the cDNAs of Gong et al. can be used in the present invention.
  • tissue-specific promoters, hormone-inducible promoters, heavy-metal inducible promoters and the like from zebrafish can be isolated and used to make fluorescent zebrafish (or other fish species) that express a GFP or variant thereof, in response to the relevant compound.
  • Multiple color fluorescent fish may be generated by the same technique as blue fluorescent protein (BFP) gene, yellow fluorescent protein (YFP) gene and cyan fluorescent protein (CFP) gene are available from Clonetech.
  • BFP blue fluorescent protein
  • YFP yellow fluorescent protein
  • CFP cyan fluorescent protein
  • a transgenic fish with GFP under an eye-specific promoter, BFP under a skin-specific promoter, and YFP under a muscle-specific promoter will show the following multiple fluorescent colors: green eyes, blue skin and yellow muscle.
  • tissue specific promoters and fluorescent protein genes By recombining different tissue specific promoters and fluorescent protein genes, more varieties of transgenic fish of different fluorescent color patterns will be created.
  • an intermediate color may be created. For example, expression of both GFP and BFP under a skin-specific promoter, a dark-green skin color may be created.
  • a biosensor system may be developed for monitoring environmental pollution and for evaluating water quality for human consumption and aquacultural uses.
  • the transgenic fish will glow with a green fluorescence (or other color depending on the fluorescence protein gene used) when pollutants such as heavy metals and estrogens (or their derivatives) reach a threshold concentration in an aquatic environment.
  • pollutants such as heavy metals and estrogens (or their derivatives) reach a threshold concentration in an aquatic environment.
  • a biosensor system has advantages over classical analytical methods because it is rapid, visualizable, and capable of identifying specific compounds directly in complex mixture found in an aquatic environment, and is portable or less instrument dependent.
  • the biosensor system also provides direct information on biotoxicity and it is biodegradable and regenerative.
  • Environmental monitoring of several substances can be accomplished by either creating one transgenic fish having genes encoding different colored fluorescent proteins driven by promoters responsive to each substance. Then the particular colors exhibited the fish in an environment can be observed. Alternatively, a number of fish can be transformed with individual vectors, then the fish can be combined into a population for monitoring an environment and the colors expressed by each fish observed.
  • the fluorescent transgenic fish should also be valuable in the market for scientific research tools because they can be used for embryonic studies such as tracing cell lineage and cell migration.
  • Cells from transgenic fish expressing GFP can also be used as cellular and genetic markers in cell transplantation and nuclear transplantation experiments.
  • the chimeric gene constructs demonstrated successfully in zebrafish in the present invention should also be applicable to other fish species such as medaka, goldfish, carp including koi, loach, tilapia, glassfish, catfish, angel fish, discus, eel, tetra, goby, gourami, guppy, Xiphophorus (swordtail), hatchet fish, Molly fish, pangasius, etc.
  • the promoters described herein can be used directly in these fish species.
  • the homologous gene promoters from other fish species can be isolated by the method described in this invention.
  • the isolated and characterized zebrafish cDNA clones and promoters described in this invention can be used as molecular probes to screen for homologous promoters in other fish species by molecular hybridization or by PCR.
  • a strong muscle-specific promoter such as MLC2f is valuable to direct a gene to be expressed in muscle tissues for generation of other beneficial transgenic fish.
  • transgenic expression of a growth hormone gene under the muscle-specific promoter may stimulate somatic growth of transgenic fish.
  • DNA can be introduced either by microinjection, electroporation, or sperm carrier to generate germ-line transgenic fish, or by direct injection of naked DNA into skeletal muscles (Xu et al., 1999) or into other tissues or cavities, or by a biolistic method (gene bombardment or gene gun) (Gomez-Chiarri et al., 1996).
  • Oligonucleotide for linker used in linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated
  • Oligonucleotide for linker used in linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated linker-mediated
  • n is a dideoxycytidine
  • GATA-1 expression pattern can be recapitulated in living transgenic zebrafish using GFP reporter gene. Development 124, 4105-4111.
  • Electroporation A method for transferring genes into the gametes of zebrafish (Brachydanio rerio), channel catfish (Ictalurus punctatus), and common carp (Cyprinus capio). Mol. Marine Biol. Biotechnol. 1, 301-308. 35. Prasher, D.C, Eckenrode, V.K., Ward, W.W., Prendergast, F.G., and Cormier, M.J. (1992). Primary structure of the Aequorea victoria green-fluorescent protein. Gene 111, 229-233.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Environmental Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Plant Pathology (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Food Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Toxicology (AREA)
  • Cell Biology (AREA)
  • Endocrinology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne quatre promoteurs géniques du poisson zèbre, spécifiques de la peau, des muscles, et des muscles du squelette, exprimés respectivement de manière ubiquiste, isolés et formant une ligature avec l'extrémité 5' du gène de la protéine fluorescente verte améliorée (EGFP). Lorsque les constructions géniques chimères résultantes sont introduites dans le poisson zèbre, le poisson zèbre transgénique émet une fluorescence verte en réponse à une lumière bleue ou ultraviolette, en fonction de la spécificité des promoteurs utilisés. Puis, on développe de nouvelles variétés de poissons d'agrément présentant différents motifs fluorescents, par exemple la fluorescence de la peau, des muscles, et la fluorescence spécifique d'un muscle du squelette et/ou ubiquiste.
PCT/SG1999/000079 1999-02-18 1999-07-16 Constructions geniques permettant de produire un poisson d'agrement transgenique fluorescent WO2000049150A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US09/913,898 US7135613B1 (en) 1999-02-18 1999-07-16 Chimeric gene constructs for generation of fluorescent transgenic ornamental fish
US10/605,708 US7834239B2 (en) 1999-02-18 2003-10-21 Sale of transgenic fish that express gene encoding fluorescent protein
US11/749,032 US8153858B2 (en) 1999-02-18 2007-05-15 Sale of fluorescent transgenic ornamental fish
US13/334,444 US8378169B2 (en) 1999-02-18 2011-12-22 Chimeric gene constructs for generation of fluorescent transgenic ornamental fish
US13/738,704 US9763430B2 (en) 1999-02-18 2013-01-10 Chimeric gene constructs for generation of fluorescent transgenic ornamental fish
US15/674,798 US11259509B2 (en) 1999-02-18 2017-08-11 Chimeric gene constructs for generation of fluorescent transgenic ornamental fish
US17/680,843 US20220256819A1 (en) 1999-02-18 2022-02-25 Chimeric gene constructs for generation of fluorescent transgenic ornamental fish

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SG1999008112 1999-02-18
SG9900811-2 1999-02-18

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US09/913,898 A-371-Of-International US7135613B1 (en) 1999-02-18 1999-07-16 Chimeric gene constructs for generation of fluorescent transgenic ornamental fish
US91389899A A-371-Of-International 1999-02-18 1999-07-16
US10/605,708 Division US7834239B2 (en) 1999-02-18 2003-10-21 Sale of transgenic fish that express gene encoding fluorescent protein

Publications (1)

Publication Number Publication Date
WO2000049150A1 true WO2000049150A1 (fr) 2000-08-24

Family

ID=59828495

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG1999/000079 WO2000049150A1 (fr) 1999-02-18 1999-07-16 Constructions geniques permettant de produire un poisson d'agrement transgenique fluorescent

Country Status (2)

Country Link
US (7) US7135613B1 (fr)
WO (1) WO2000049150A1 (fr)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1180684A1 (fr) * 2000-08-17 2002-02-20 President of Hiroshima University Poisson médaka hautement sensible aux oestrogènes
WO2002088368A1 (fr) * 2001-05-02 2002-11-07 Institute Of Molecular Agrobiology Regulation spatiale et temporelle au moyen d'un baculovirus de l'expression de genes chez le poisson zebre
EP1497643A1 (fr) * 2002-04-09 2005-01-19 F.C. Thomas Allnut Systemes d'aquacultures clos destines a produire des proteines recombinees purifiees
EP1571222A2 (fr) * 2004-03-03 2005-09-07 Taikong Corporation Fragments génétiques fluorescents et poissons transgéniques correspondants
CN1297664C (zh) * 2003-08-29 2007-01-31 邰港科技股份有限公司 表达二荧光基因之重组质粒
CN102128938A (zh) * 2010-12-20 2011-07-20 长春理工大学 甾体类激素及多环芳烃高效生物荧光传感器及构建方法
US8232451B1 (en) 2011-02-15 2012-07-31 Yorktown Technologies, L.P. Blue transgenic fluorescent ornamental fish
US8232450B1 (en) 2011-02-15 2012-07-31 Yorktown Technologies, L.P. Purple transgenic fluorescent ornamental fish
CN102911937A (zh) * 2011-08-03 2013-02-06 龚纮毅 肌肉增强子序列及其应用
TWI396742B (zh) * 2009-10-30 2013-05-21 Academia Sinica 新穎啟動子片段及其應用
CN103540611A (zh) * 2013-09-26 2014-01-29 马明 一种生产可变色光诱导荧光观赏鱼的方法
EP2703413A1 (fr) * 2012-08-30 2014-03-05 Body Organ Biomedical Corp. Vecteur de recombinaison, oeuf de poisson transgénique utilisant celui-ci et biomatériau l'utilisant
US8727554B2 (en) 2007-06-25 2014-05-20 Yorktown Technologies, L.P. Aquarium with adjustable lighting
US8975467B2 (en) 2012-06-12 2015-03-10 Yorktown Technologies, L.P. Green transgenic fluorescent ornamental fish
US8987546B2 (en) 2012-11-06 2015-03-24 Yorktown Technologies, L.P. Orange transgenic fluorescent ornamental fish
US9282729B2 (en) 2014-01-31 2016-03-15 Yorktown Technologies, L.P. Purple transgenic fluorescent ornamental fish
US9295238B2 (en) 2014-01-31 2016-03-29 Yorktown Technologies, L.P. Pink transgenic fluorescent ornamental fish
US9363986B2 (en) 2014-01-31 2016-06-14 Yorktown Technologies, L.P. Orange transgenic fluorescent ornamental fish
US9380768B2 (en) 2014-01-31 2016-07-05 Yorktown Technologies, L.P. Red transgenic fluorescent ornamental fish
US9392776B2 (en) 2012-07-26 2016-07-19 Yorktown Technologies, L.P. Green transgenic fluorescent ornamental fish
CN106577416A (zh) * 2016-12-08 2017-04-26 金华职业技术学院 一种基于荧光蛋白的夜明珠培育方法
US9968077B2 (en) 2014-11-07 2018-05-15 Glofish Llc Blue transgenic fluorescent ornamental fish
CN110684777A (zh) * 2019-11-13 2020-01-14 华中农业大学 一段分离的核苷酸序列在肌间刺减少的斑马鱼构建中的应用
WO2023229363A1 (fr) * 2022-05-27 2023-11-30 고려대학교 산학협력단 Modèle animal de sclérose latérale amyotrophique utilisant le poisson-zèbre transgénique et sa méthode de construction

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070071776A1 (en) * 1998-09-28 2007-03-29 National University Of Singapore Recombinant Vaccine Against Fish Infectious Diseases
US7700825B2 (en) 2006-08-16 2010-04-20 Yorktown Technologies, L.P. Recombinant constructs and transgenic fluorescent ornamental fish therefrom
US20090006219A1 (en) * 2007-06-29 2009-01-01 Alan Blake Method for distribution of an aquarium kit
KR101255197B1 (ko) * 2010-06-28 2013-04-23 부경대학교 산학협력단 더블 형질전환 불임성 형광 바다 송사리
CN102147364B (zh) * 2010-12-20 2012-12-05 长春理工大学 一种检测甾体类激素及多环芳烃的高效微生物荧光传感器
TWI402343B (zh) * 2011-01-20 2013-07-21 Univ Nat Taiwan Ocean 新穎肌肉增強子序列及其應用
US9534235B2 (en) 2011-02-01 2017-01-03 Purdue Research Foundation Efficient sterilization of fish by disruption of germ cell development
US20130031644A1 (en) * 2011-05-23 2013-01-31 University Of Tennessee Research Foundation Autonomous lux reporter system and methods of use
US10342223B2 (en) 2013-02-13 2019-07-09 GloFish, LLC Mollusk with coated shell
US10098334B2 (en) 2016-03-11 2018-10-16 GloFish, LLC Red transgenic fluorescent ornamental fish
CA3057141A1 (fr) * 2017-03-30 2018-10-04 GloFish, LLC Requin arc-en-ciel transgenique
US10346783B2 (en) * 2017-09-14 2019-07-09 Baker Hughes, A Ge Company, Llc Electrochemical methods of removing dissolved oxygen from drilling or completion fluids
GB2595137A (en) * 2019-01-10 2021-11-17 Glofish Llc Transgenic barb
CN112430624B (zh) * 2020-11-25 2023-08-18 汕头大学 一种斑马鱼肌肉特异诱导型表达载体及应用
TW202241260A (zh) * 2021-01-12 2022-11-01 美商螢光魚有限責任公司 基因轉殖兵鯰

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996003034A1 (fr) * 1994-07-27 1996-02-08 Massachusetts Institute Of Technology Procede d'insertion de vecteur retroviral chez les poissons
WO1998015627A1 (fr) * 1996-10-10 1998-04-16 University Of Southampton Production d'un peptide recombinant par un poisson transgenique
WO1998056902A2 (fr) * 1997-06-09 1998-12-17 Medical College Of Georgia Research Institute, Inc. Poissons transgeniques a expression a specificite cellulaire

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8823691D0 (en) * 1988-10-08 1988-11-16 Emi Plc Thorn Aquarium lighting
US5998698A (en) 1995-06-07 1999-12-07 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Transgenic fish capable of expressing exogenous lytic peptides
US5876995A (en) 1996-02-06 1999-03-02 Bryan; Bruce Bioluminescent novelty items
US6247995B1 (en) 1996-02-06 2001-06-19 Bruce Bryan Bioluminescent novelty items
DE69938293T2 (de) * 1998-03-27 2009-03-12 Bruce J. Beverly Hills Bryan Luciferase, gfp fluoreszenzproteine, kodierende nukleinsaüre und ihre verwendung in der diagnose
DE69926629T2 (de) 1998-05-31 2006-06-08 University Of Georgia Research Foundation, Inc. Transgene fische, die ein von bakteriophagen abgeleitetes transgenes konstrukt zum nachweis von mutationen tragen
US20020013955A1 (en) 1998-06-10 2002-01-31 Sharon Ogden Production of recombinant protein in transgenic fish
WO2000024246A2 (fr) 1998-10-26 2000-05-04 The University Of Georgia Research Foundation, Inc. Poisson transgenique a base de plasmide permettant de detecter une mutation et techniques

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996003034A1 (fr) * 1994-07-27 1996-02-08 Massachusetts Institute Of Technology Procede d'insertion de vecteur retroviral chez les poissons
WO1998015627A1 (fr) * 1996-10-10 1998-04-16 University Of Southampton Production d'un peptide recombinant par un poisson transgenique
WO1998056902A2 (fr) * 1997-06-09 1998-12-17 Medical College Of Georgia Research Institute, Inc. Poissons transgeniques a expression a specificite cellulaire

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 127, no. 10, 8 September 1997, Columbus, Ohio, US; abstract no. 131871Y, MULLER ET AL.: "Activator effect of coinjected enhancers on the muscle-specific expression of promoters of zebrafish embryos" page 195; *
MOL. REPROD.,, vol. 47, no. 4, December 1997 (1997-12-01), (ENG), pages 404 - 412 *

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1180684A1 (fr) * 2000-08-17 2002-02-20 President of Hiroshima University Poisson médaka hautement sensible aux oestrogènes
US6759568B2 (en) 2000-08-17 2004-07-06 President Of Hiroshima University High estrogen-sensitive medaka fish
WO2002088368A1 (fr) * 2001-05-02 2002-11-07 Institute Of Molecular Agrobiology Regulation spatiale et temporelle au moyen d'un baculovirus de l'expression de genes chez le poisson zebre
EP1497643A1 (fr) * 2002-04-09 2005-01-19 F.C. Thomas Allnut Systemes d'aquacultures clos destines a produire des proteines recombinees purifiees
EP1497643A4 (fr) * 2002-04-09 2008-07-30 F C Thomas Allnut Systemes d'aquacultures clos destines a produire des proteines recombinees purifiees
CN1297664C (zh) * 2003-08-29 2007-01-31 邰港科技股份有限公司 表达二荧光基因之重组质粒
EP1571222A3 (fr) * 2004-03-03 2007-05-09 Taikong Corporation Fragments génétiques fluorescents et poissons transgéniques correspondants
EP1571222A2 (fr) * 2004-03-03 2005-09-07 Taikong Corporation Fragments génétiques fluorescents et poissons transgéniques correspondants
US9374987B1 (en) 2007-06-25 2016-06-28 Yorktown Technologies, L.P. Aquarium with adjustable lighting
US8727554B2 (en) 2007-06-25 2014-05-20 Yorktown Technologies, L.P. Aquarium with adjustable lighting
US9295236B2 (en) 2007-06-25 2016-03-29 Yorktown Technologies, L.P. Aquarium with adjustable lighting
USRE49345E1 (en) 2007-06-25 2022-12-27 GloFish, LLC Aquarium with adjustable lighting
USRE48169E1 (en) 2007-06-25 2020-08-25 Glofish Llc Aquarium with adjustable lighting
TWI396742B (zh) * 2009-10-30 2013-05-21 Academia Sinica 新穎啟動子片段及其應用
CN102128938B (zh) * 2010-12-20 2013-06-19 长春理工大学 甾体类激素及多环芳烃高效生物荧光传感器及构建方法
CN102128938A (zh) * 2010-12-20 2011-07-20 长春理工大学 甾体类激素及多环芳烃高效生物荧光传感器及构建方法
US8232450B1 (en) 2011-02-15 2012-07-31 Yorktown Technologies, L.P. Purple transgenic fluorescent ornamental fish
US8581024B2 (en) 2011-02-15 2013-11-12 Yorktown Technologies, L.P. Blue transgenic fluorescent ornamental fish
US8581023B2 (en) 2011-02-15 2013-11-12 Yorktown Technologies, L.P. Purple transgenic fluorescent ornamental fish
US8232451B1 (en) 2011-02-15 2012-07-31 Yorktown Technologies, L.P. Blue transgenic fluorescent ornamental fish
CN102911937B (zh) * 2011-08-03 2014-03-26 龚纮毅 肌肉增强子序列及其应用
CN102911937A (zh) * 2011-08-03 2013-02-06 龚纮毅 肌肉增强子序列及其应用
US8975467B2 (en) 2012-06-12 2015-03-10 Yorktown Technologies, L.P. Green transgenic fluorescent ornamental fish
US9392776B2 (en) 2012-07-26 2016-07-19 Yorktown Technologies, L.P. Green transgenic fluorescent ornamental fish
EP2703413A1 (fr) * 2012-08-30 2014-03-05 Body Organ Biomedical Corp. Vecteur de recombinaison, oeuf de poisson transgénique utilisant celui-ci et biomatériau l'utilisant
US8987546B2 (en) 2012-11-06 2015-03-24 Yorktown Technologies, L.P. Orange transgenic fluorescent ornamental fish
CN103540611A (zh) * 2013-09-26 2014-01-29 马明 一种生产可变色光诱导荧光观赏鱼的方法
US9282729B2 (en) 2014-01-31 2016-03-15 Yorktown Technologies, L.P. Purple transgenic fluorescent ornamental fish
US9295238B2 (en) 2014-01-31 2016-03-29 Yorktown Technologies, L.P. Pink transgenic fluorescent ornamental fish
US9363986B2 (en) 2014-01-31 2016-06-14 Yorktown Technologies, L.P. Orange transgenic fluorescent ornamental fish
US9380768B2 (en) 2014-01-31 2016-07-05 Yorktown Technologies, L.P. Red transgenic fluorescent ornamental fish
US10798923B2 (en) 2014-11-07 2020-10-13 GloFish, LLC Blue transgenic fluorescent ornamental fish
US9968077B2 (en) 2014-11-07 2018-05-15 Glofish Llc Blue transgenic fluorescent ornamental fish
US11716975B2 (en) 2014-11-07 2023-08-08 Glofish Llc Blue transgenic fluorescent ornamental fish
CN106577416A (zh) * 2016-12-08 2017-04-26 金华职业技术学院 一种基于荧光蛋白的夜明珠培育方法
CN110684777A (zh) * 2019-11-13 2020-01-14 华中农业大学 一段分离的核苷酸序列在肌间刺减少的斑马鱼构建中的应用
CN110684777B (zh) * 2019-11-13 2021-05-07 华中农业大学 一段分离的核苷酸序列在肌间刺减少的斑马鱼构建中的应用
WO2023229363A1 (fr) * 2022-05-27 2023-11-30 고려대학교 산학협력단 Modèle animal de sclérose latérale amyotrophique utilisant le poisson-zèbre transgénique et sa méthode de construction

Also Published As

Publication number Publication date
US20080052787A1 (en) 2008-02-28
US7834239B2 (en) 2010-11-16
US8153858B2 (en) 2012-04-10
US20120167241A1 (en) 2012-06-28
US11259509B2 (en) 2022-03-01
US8378169B2 (en) 2013-02-19
US7135613B1 (en) 2006-11-14
US20180064074A1 (en) 2018-03-08
US20040143864A1 (en) 2004-07-22
US9763430B2 (en) 2017-09-19
US20220256819A1 (en) 2022-08-18
US20140007265A1 (en) 2014-01-02

Similar Documents

Publication Publication Date Title
US20220256819A1 (en) Chimeric gene constructs for generation of fluorescent transgenic ornamental fish
US5545808A (en) Transgenic salmonid fish expressing exogenous salmonid growth hormone
EP0986640B1 (fr) Poissons transgeniques a expression a specificite cellulaire
Amsterdam et al. The Aequorea victoria green fluorescent protein can be used as a reporter in live zebrafish embryos
Chou et al. Uniform GFP-expression in transgenic medaka (Oryzias latipes) at the F0 generation
Cho et al. Functional ability of cytoskeletal β-actin regulator to drive constitutive and ubiquitous expression of a fluorescent reporter throughout the life cycle of transgenic marine medaka Oryzias dancena
Takeuchi et al. Green fluorescent protein as a cell-labeling tool and a reporter of gene expression in transgenic rainbow trout
WO2004023867A2 (fr) Modelisation du cancer a l'aide de poissons transgeniques
US20040110225A1 (en) Fluorescent proteins from aquatic species
US20060242719A1 (en) Nucleotide sequences of shrimp beta-actin and actin promoters and their use in gentic transformation technology
US7427677B2 (en) Expression of zebrafish bone morphogenetic protein 4
US20040216179A1 (en) Recombinant plasmid expressing two fluorescence genes
RU2807599C2 (ru) Генетически модифицированные стерильные птицы и способ их воспроизводства
Yazawa et al. Promoter analysis of several tissue-specific and inducible Japanese flounder, Paralichtys olivaceus genes
JPH09508526A (ja) 軟体動物の遺伝子工学
Chiou et al. 22 Application of Transgenic Fish Technology in Sea Food Production

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 09913898

Country of ref document: US

122 Ep: pct application non-entry in european phase